Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 461
Filter
1.
Nature ; 626(8001): 1084-1093, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38355799

ABSTRACT

The house mouse (Mus musculus) is an exceptional model system, combining genetic tractability with close evolutionary affinity to humans1,2. Mouse gestation lasts only 3 weeks, during which the genome orchestrates the astonishing transformation of a single-cell zygote into a free-living pup composed of more than 500 million cells. Here, to establish a global framework for exploring mammalian development, we applied optimized single-cell combinatorial indexing3 to profile the transcriptional states of 12.4 million nuclei from 83 embryos, precisely staged at 2- to 6-hour intervals spanning late gastrulation (embryonic day 8) to birth (postnatal day 0). From these data, we annotate hundreds of cell types and explore the ontogenesis of the posterior embryo during somitogenesis and of kidney, mesenchyme, retina and early neurons. We leverage the temporal resolution and sampling depth of these whole-embryo snapshots, together with published data4-8 from earlier timepoints, to construct a rooted tree of cell-type relationships that spans the entirety of prenatal development, from zygote to birth. Throughout this tree, we systematically nominate genes encoding transcription factors and other proteins as candidate drivers of the in vivo differentiation of hundreds of cell types. Remarkably, the most marked temporal shifts in cell states are observed within one hour of birth and presumably underlie the massive physiological adaptations that must accompany the successful transition of a mammalian fetus to life outside the womb.


Subject(s)
Animals, Newborn , Embryo, Mammalian , Embryonic Development , Gastrula , Single-Cell Analysis , Time-Lapse Imaging , Animals , Female , Mice , Pregnancy , Animals, Newborn/embryology , Animals, Newborn/genetics , Cell Differentiation/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryonic Development/genetics , Gastrula/cytology , Gastrula/embryology , Gastrulation/genetics , Kidney/cytology , Kidney/embryology , Mesoderm/cytology , Mesoderm/enzymology , Neurons/cytology , Neurons/metabolism , Retina/cytology , Retina/embryology , Somites/cytology , Somites/embryology , Time Factors , Transcription Factors/genetics , Transcription, Genetic , Organ Specificity/genetics
2.
Elife ; 112022 01 20.
Article in English | MEDLINE | ID: mdl-35049495

ABSTRACT

Genomic imprinting refers to the mono-allelic and parent-specific expression of a subset of genes. While long recognized for their role in embryonic development, imprinted genes have recently emerged as important modulators of postnatal physiology, notably through hypothalamus-driven functions. Here, using mouse models of loss, gain and parental inversion of expression, we report that the paternally expressed Zdbf2 gene controls neonatal growth in mice, in a dose-sensitive but parent-of-origin-independent manner. We further found that Zdbf2-KO neonates failed to fully activate hypothalamic circuits that stimulate appetite, and suffered milk deprivation and diminished circulating Insulin Growth Factor 1 (IGF-1). Consequently, only half of Zdbf2-KO pups survived the first days after birth and those surviving were smaller. This study demonstrates that precise imprinted gene dosage is essential for vital physiological functions at the transition from intra- to extra-uterine life, here the adaptation to oral feeding and optimized body weight gain.


Subject(s)
DNA-Binding Proteins/genetics , Eating/genetics , Genomic Imprinting/genetics , Hypothalamus , Weight Gain/genetics , Animals , Animals, Newborn/genetics , Animals, Newborn/physiology , Female , Hypothalamus/metabolism , Hypothalamus/physiology , Male , Mice , Mice, Knockout , Pregnancy
3.
Genes (Basel) ; 12(12)2021 11 25.
Article in English | MEDLINE | ID: mdl-34946830

ABSTRACT

Selenium (Se) is an essential micronutrient for growth and immune function in beef cattle. We previously showed that supranutritional maternal organic Se supplementation during late pregnancy improves immune function in their newborn calves; however, the effects of maternal organic Se-supplementation on fetal programming during different pregnancy stages have yet to be elucidated. Herein, we investigated the effects of supranutritional maternal organic Se-supplementation in different pregnancy trimesters on their beef calf's genome-wide transcriptome profiles. Within 12 to 48 h of birth, whole blood and Longissimus dorsi (LD) muscle biopsies were collected from calves born to 40 crossbred Angus cows that received, except for the control group (CTR), Se-yeast boluses (105 mg of Se/wk) during the first (TR1), second (TR2), or third (TR3) trimester of gestation. Whole-blood Se concentrations of newborn calves increased from CTR, TR1, TR2 to TR3, whereas muscle Se concentrations of newborn calves were only increased in TR3 group. We identified 3048 unique differentially expressed genes (DEGs) across all group comparisons (FDR ≤ 0.05 and |log2FC| ≥ 1.5). Furthermore, we predicted 237 unique transcription factors that putatively regulate the DEGs. Independent of supplementation trimester, supranutritional maternal organic Se supplementation downregulated genes involved in adaptive immunity in all trimesters. Dependent on supplementation trimester, genes involved in muscle development were upregulated by TR3 Se supplementation and downregulated by TR1 Se-supplementation, and genes involved in collagen formation were downregulated by TR2 Se-supplementation. Supranutritional maternal organic Se supplementation in the last trimester of pregnancy resulted in upregulation of myosin and actin filament associated genes, potentially allowing for optimal muscle function and contraction. Our findings suggest a beneficial effect of supranutritional maternal organic Se supplementation during late gestation on Se-status and muscle development and function of newborn calves.


Subject(s)
Muscles/metabolism , Pregnancy Trimesters/drug effects , Selenium/administration & dosage , Transcriptome/drug effects , Transcriptome/genetics , Animal Feed , Animals , Animals, Newborn/genetics , Cattle , Dietary Supplements , Female , Parturition/genetics , Pregnancy , Pregnancy Trimesters/genetics
4.
FASEB J ; 35(9): e21806, 2021 09.
Article in English | MEDLINE | ID: mdl-34369605

ABSTRACT

During lactation, adult female mice display aggressive responses toward male intruders, triggered by male-derived chemosensory signals. This aggressive behavior is not shown by pup-sensitized virgin females sharing pup care with dams. The genetic mechanisms underlying the switch from attraction to aggression are unknown. In this work, we investigate the differential gene expression in lactating females expressing maternal aggression compared to pup-sensitized virgin females in the medial amygdala (Me), a key neural structure integrating chemosensory and hormonal information. The results showed 197 genes upregulated in dams, including genes encoding hormones such as prolactin, growth hormone, or follicle-stimulating hormone, neuropeptides such as galanin, oxytocin, and pro-opiomelanocortin, and genes related to catecholaminergic and cholinergic neurotransmission. In contrast, 99 genes were downregulated in dams, among which we find those encoding for inhibins and transcription factors of the Fos and early growth response families. The gene set analysis revealed numerous Gene Ontology functional groups with higher expression in dams than in pup-sensitized virgin females, including those related with the regulation of the Jak/Stat cascade. Of note, a number of olfactory and vomeronasal receptor genes was expressed in the Me, although without differences between dams and virgins. For prolactin and growth hormone, a qPCR experiment comparing dams, pup-sensitized, and pup-naïve virgin females showed that dams expressed higher levels of both hormones than pup-naïve virgins, with pup-sensitized virgins showing intermediate levels. Altogether, the results show important gene expression changes in the Me, which may underlie some of the behavioral responses characterizing maternal behavior.


Subject(s)
Amygdala/physiology , Animals, Newborn/genetics , Gene Expression/genetics , Lactation/genetics , Maternal Behavior/physiology , Animals , Female , Hormones/genetics , Mice , Models, Animal , Pregnancy , Receptors, Odorant/genetics , Vomeronasal Organ/physiology
5.
Nature ; 596(7870): 92-96, 2021 08.
Article in English | MEDLINE | ID: mdl-34321664

ABSTRACT

The mammalian brain develops through a complex interplay of spatial cues generated by diffusible morphogens, cell-cell interactions and intrinsic genetic programs that result in probably more than a thousand distinct cell types. A complete understanding of this process requires a systematic characterization of cell states over the entire spatiotemporal range of brain development. The ability of single-cell RNA sequencing and spatial transcriptomics to reveal the molecular heterogeneity of complex tissues has therefore been particularly powerful in the nervous system. Previous studies have explored development in specific brain regions1-8, the whole adult brain9 and even entire embryos10. Here we report a comprehensive single-cell transcriptomic atlas of the embryonic mouse brain between gastrulation and birth. We identified almost eight hundred cellular states that describe a developmental program for the functional elements of the brain and its enclosing membranes, including the early neuroepithelium, region-specific secondary organizers, and both neurogenic and gliogenic progenitors. We also used in situ mRNA sequencing to map the spatial expression patterns of key developmental genes. Integrating the in situ data with our single-cell clusters revealed the precise spatial organization of neural progenitors during the patterning of the nervous system.


Subject(s)
Brain/cytology , Brain/embryology , Single-Cell Analysis , Transcriptome , Animals , Animals, Newborn/genetics , Brain/anatomy & histology , Female , Gastrulation/genetics , Male , Mice , Neural Tube/anatomy & histology , Neural Tube/cytology , Neural Tube/embryology
6.
Biol Reprod ; 105(3): 720-734, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34258621

ABSTRACT

Each year, millions of infants and children are anesthetized for medical and surgical procedures. Yet, a substantial body of preclinical evidence suggests that anesthetics are neurotoxins that cause rapid and widespread apoptotic cell death in the brains of infant rodents and nonhuman primates. These animals have persistent impairments in cognition and behavior many weeks or months after anesthesia exposure, leading us to hypothesize that anesthetics do more than simply kill brain cells. Indeed, anesthetics cause chronic neuropathology in neurons that survive the insult, which then interferes with major aspects of brain development, synaptic plasticity, and neuronal function. Understanding the phenomenon of anesthesia-induced developmental neurotoxicity is of critical public health importance because clinical studies now report that anesthesia in human infancy is associated with cognitive and behavioral deficits. In our search for mechanistic explanations for why a young and pliable brain cannot fully recover from a relatively brief period of anesthesia, we have accumulated evidence that neonatal anesthesia can dysregulate epigenetic tags that influence gene transcription such as histone acetylation and DNA methylation. In this review, we briefly summarize the phenomenon of anesthesia-induced developmental neurotoxicity. We then discuss chronic neuropathology caused by neonatal anesthesia, including disturbances in cognition, socio-affective behavior, neuronal morphology, and synaptic plasticity. Finally, we present evidence of anesthesia-induced genetic and epigenetic dysregulation within the developing brain that may be transmitted intergenerationally to anesthesia-naïve offspring.


Subject(s)
Anesthesia/adverse effects , Animals, Newborn/genetics , Epigenome/drug effects , Primates/genetics , Animals , Humans , Infant, Newborn , Mice , Rats
7.
Radiat Res ; 196(2): 183-191, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34019665

ABSTRACT

Ionizing radiation exposure is associated with a risk of cardiac fibrosis; however, the underlying molecular mechanism remains unclear. Growth/differentiation factor-15 (GDF15), a fibroblast factor, is a divergent member of the transforming growth factor ß superfamily. Next-generation sequencing analyses has revealed that Gdf15 is increased in cardiac fibroblasts during radiation-induced fibrosis. However, the role of Gdf15 in cardiac fibrosis remains unclear. In this study, we demonstrated that the upregulated expression of GDF15 in newborn rat cardiac fibroblasts and adult rats after irradiation could induce fibrosis, which was confirmed by the increased cell proliferation rate and the increased expression of fibrosis markers (Col1α and αSMA) in newborn rat cardiac fibroblasts after transfection with Gdf15 in vitro. Conversely, the downregulation of GDF15 inhibited cardiac fibrosis, as confirmed by G2/M-cell cycle arrest, suppression of cell proliferation, and low levels of Col1α and αSMA expression. We also found that suppressing the expression of Gdf15 in cardiac fibroblasts could lead to a decrease in CDK1 and inhibit phosphorylation of ERK1/2. Thus, GDF15 might promote cardiac fibroblast fibrosis through the MAPK/ERK1/2 pathway and thus contribute to the pathogenesis of radiation-induced heart disease.


Subject(s)
Fibrosis/genetics , Growth Differentiation Factor 15/genetics , Heart/radiation effects , Radiation, Ionizing , Actins/genetics , Animals , Animals, Newborn/genetics , Cell Proliferation/radiation effects , Collagen Type I/genetics , Collagen Type I, alpha 1 Chain , Fibroblasts/radiation effects , Fibrosis/etiology , Fibrosis/pathology , Gene Expression Regulation/radiation effects , Heart/physiopathology , Humans , Mitogen-Activated Protein Kinase Kinases/genetics , Rats , Signal Transduction/radiation effects
8.
J Pharmacol Sci ; 146(2): 105-115, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33941321

ABSTRACT

OBJECTIVE: To investigate the effect of sevoflurane on inflammation of microglia in hippocampus of neonatal rats, and to investigate whether the related mechanism is related to Wnt/ß-Catenin/CaMKIV pathway. METHODS: Neonatal rats were anesthetized with 2% or 3% sevoflurane for 4 h a day for 3 consecutive days. Water maze test was used to detect the effect of sevoflurane anesthesia on memory function of neonatal rats. H&E and Nissl staining were used to observe the pathological damage of hippocampal area of neonatal rats induced by sevoflurane anesthesia. The expression of microglial marker Iba-1 was detected by Immunofluorescence. Immunofluorescence and WB were used to detect the expression CD32b, CD86, TNF-α, IL-6, Wnt3a, ß-Catenin and CaMKIV in hippocampus. To further explore the related mechanism, Wnt-3α inhibitor and activator was treated to study the effect of sevoflurane on microglial inflammation in hippocampus of neonatal rats. RESULTS: Sevoflurane anesthesia significantly increased escape latency time, reduced platform crossing times, and damaged the learning and memory ability of neonatal rats. H&E and Nissl staining results showed that sevoflurane anesthesia caused obvious damage to the hippocampus of neonatal rats. Sevoflurane anesthesia promoted the expression of Iba-1 and activated microglia. Sevoflurane anesthesia not only significantly increased the positive expression of CD32b, CD86, TNF-α and IL-6, but also decreased the expression of Wnt3a, ß-Catenin and CaMKIV. These results suggested that sevoflurane inhibited Wnt/ß-Catenin/CaMKIV pathway. CONCLUSION: Sevoflurane induces inflammation of microglia in hippocampus of neonatal rats by inhibiting Wnt/ß-Catenin/CaMKIV pathway.


Subject(s)
Anesthetics, Inhalation/adverse effects , Animals, Newborn/genetics , Animals, Newborn/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 4/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Inflammation/etiology , Microglia/metabolism , Microglia/pathology , Sevoflurane/adverse effects , Signal Transduction/drug effects , Signal Transduction/genetics , Wnt Proteins/metabolism , beta Catenin/metabolism , Animals , Gene Expression/drug effects , Inflammation/genetics , Inflammation/metabolism , Inflammation Mediators/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Male , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
9.
PLoS One ; 16(4): e0242635, 2021.
Article in English | MEDLINE | ID: mdl-33901192

ABSTRACT

Plasma cell-free DNA (cfDNA) levels have been associated with disease and survival status in septic humans and dogs. To date, studies investigating cfDNA levels in association with critical illness in foals are lacking. We hypothesized that cfDNA would be detectable in the plasma of foals, that septic and sick-nonseptic foals would have significantly higher cfDNA levels compared to healthy foals, and that increased cfDNA levels would be associated with non-survival. Animals used include 80 foals of 10 days of age or less admitted to a tertiary referral center between January and July, 2020 were stratified into three categories: healthy (n = 34), sick non-septic (n = 11) and septic (n = 35) based on specific criteria. This was a prospective clinical study. Blood was collected from critically ill foals at admission or born in hospital for cfDNA quantification and blood culture. Previously published sepsis score (SS) and neonatal SIRS score (NSIRS) were also calculated. SS, NSIRS, blood culture status and cfDNA concentrations were evaluated to predict survival. Continuous variables between groups were compared using Kruskal-Wallis ANOVA with Dunn's post hoc test. Comparisons between two groups were assessed using the Mann-Whitney U-test or Spearman rank for correlations. The performance of cfDNA, sepsis score and NSIRS score to predict survival was assessed by receiver operator characteristic (ROC) curve analysis including area under the curve, sensitivity and specificity using cutoffs. Plasma cfDNA was detectable in all foals. No significant differences in cfDNA concentration were detected between healthy foals and septic foals (P = 0.65) or healthy foals and sick non-septic foals (P = 0.88). There was no significant association between cfDNA and culture status, SS, NSIRS or foal survival. SS (AUC 0.85) and NSIRS (AUC 0.83) were superior to cfDNA (AUC 0.64) in predicting survival. Although cfDNA was detectable in foal plasma, it offers negligible utility to diagnose sepsis or predict survival in critical illness in neonates.


Subject(s)
Animals, Newborn/genetics , Cell-Free Nucleic Acids/genetics , Critical Illness/mortality , Horse Diseases/genetics , Horse Diseases/mortality , Horses/genetics , Animals , Prospective Studies , ROC Curve , Sensitivity and Specificity , Sepsis/genetics , Sepsis/mortality , Sepsis/veterinary , Severity of Illness Index , Statistics, Nonparametric , Systemic Inflammatory Response Syndrome/genetics , Systemic Inflammatory Response Syndrome/mortality , Systemic Inflammatory Response Syndrome/veterinary
10.
Proc Natl Acad Sci U S A ; 118(18)2021 05 04.
Article in English | MEDLINE | ID: mdl-33903237

ABSTRACT

Precise spatiotemporal control of gene expression in the developing brain is critical for neural circuit formation, and comprehensive expression mapping in the developing primate brain is crucial to understand brain function in health and disease. Here, we developed an unbiased, automated, large-scale, cellular-resolution in situ hybridization (ISH)-based gene expression profiling system (GePS) and companion analysis to reveal gene expression patterns in the neonatal New World marmoset cortex, thalamus, and striatum that are distinct from those in mice. Gene-ontology analysis of marmoset-specific genes revealed associations with catalytic activity in the visual cortex and neuropsychiatric disorders in the thalamus. Cortically expressed genes with clear area boundaries were used in a three-dimensional cortical surface mapping algorithm to delineate higher-order cortical areas not evident in two-dimensional ISH data. GePS provides a powerful platform to elucidate the molecular mechanisms underlying primate neurobiology and developmental psychiatric and neurological disorders.


Subject(s)
Brain/metabolism , Callithrix/genetics , Transcriptome/genetics , Animals , Animals, Newborn/genetics , Animals, Newborn/growth & development , Brain/growth & development , Callithrix/growth & development , Corpus Striatum/growth & development , Corpus Striatum/metabolism , Gene Expression Regulation, Developmental/genetics , Humans , In Situ Hybridization , Mice , Species Specificity , Visual Cortex/growth & development , Visual Cortex/metabolism
11.
J Cell Physiol ; 236(2): 1281-1294, 2021 02.
Article in English | MEDLINE | ID: mdl-32654195

ABSTRACT

Cardiomyocyte migration represents a requisite event of cardiogenesis and the regenerative response of the injured adult zebrafish and neonatal rodent heart. The present study tested the hypothesis that the appearance of the intermediate filament protein nestin in neonatal rat ventricular cardiomyocytes (NNVMs) was associated in part with the acquisition of a migratory phenotype. The cotreatment of NNVMs with phorbol 12,13-dibutyrate (PDBu) and the p38α/ß mitogen-activated protein kinase inhibitor SB203580 led to the de novo synthesis of nestin. The intermediate filament protein was subsequently reorganized into a filamentous pattern and redistributed to the leading edge of elongated membrane protrusions translating to significant lengthening of NNVMs. PDBu/SB203580 treatment concomitantly promoted the reorganization of nonmuscle myosin IIB (NMIIB) located predominantly at the periphery of the plasma membrane of NNVMs to a filamentous phenotype extending to the leading edge of elongated membrane protrusions. Coimmunoprecipitation assay revealed a physical interaction between NMIIB and nestin after PDBu/SB203580 treatment of NNVMs. In wild-type and heterozygous NMIIB embryonic hearts at E11.5-E14.5 days, nestin immunoreactivity was identified in a subpopulation of cardiomyocytes elongating perpendicular to the compact myocardium, at the leading edge of nascent trabeculae and during thickening of the compact myocardium. In mutant embryonic hearts lacking NMIIB protein expression, trabeculae formation was reduced, the compact myocardium significantly thinner and nestin immunoreactivity undetectable in cardiomyocytes at E14.5 days. These data suggest that NMIIB and nestin may act in a coordinated fashion to facilitate the acquisition of a migratory phenotype in neonatal and embryonic cardiomyocytes.


Subject(s)
Heart/growth & development , Mitogen-Activated Protein Kinase 14/genetics , Nestin/biosynthesis , Nonmuscle Myosin Type IIB/genetics , Organogenesis/genetics , Animals , Animals, Newborn/genetics , Animals, Newborn/growth & development , Cell Movement/drug effects , Cell Proliferation/drug effects , Cytoskeleton/drug effects , Cytoskeleton/genetics , Gene Expression Regulation, Developmental/genetics , Heart/drug effects , Heart Ventricles/drug effects , Heart Ventricles/growth & development , Humans , Imidazoles/pharmacology , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Nestin/genetics , Phorbol 12,13-Dibutyrate/pharmacology , Pyridines/pharmacology , Rats , Zebrafish/genetics
12.
Transgenic Res ; 29(5-6): 553-562, 2020 12.
Article in English | MEDLINE | ID: mdl-33184751

ABSTRACT

Disco-interacting protein 2 is a highly conserved three-domain protein with two tandem Adenylate-forming domains. It is proposed to influence the processes involved in neuronal development by influencing lipid metabolism and remains to be characterized. In this study, we show that Disco-interacting protein 2a null mice do not exhibit overt phenotype defects. However, the body composition differences were observed in these mice under different dietary regimens. The neutral lipid composition of two different diets was characterized, and it was observed that the new-born mice grow relatively slower than the wild-type mice with delayed appearance of features such as dentition when fed with high-triacylglycerol NIN-formulation diet. The high-diacylglycerol Safe-formulation diet was found to accumulate more fat mass in mice than those fed with high-triacylglycerol NIN-formulation diet beyond 10 months. These findings point to a proposed relationship between dietary components (particularly the lipid composition) and body composition along with the growth of neonates in mice lacking the gene Disco-interacting protein 2a.


Subject(s)
Animals, Newborn/growth & development , Nuclear Proteins/genetics , Obesity/genetics , Adipose Tissue/physiopathology , Animal Feed , Animals , Animals, Newborn/genetics , Body Composition/genetics , Diet/adverse effects , Diglycerides/pharmacology , Female , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/metabolism , Obesity/etiology , Triglycerides/pharmacology
13.
Exp Mol Pathol ; 117: 104551, 2020 12.
Article in English | MEDLINE | ID: mdl-33010296

ABSTRACT

Cellular senescence is important for the maintenance of tissue homeostasis during normal development. In this study, we aimed to investigate the effect of renin angiotensin system (RAS) blockade on renal cell senescence in the developing rat kidney. Newborn rat pups were treated with enalapril (30 mg/kg/day) or vehicle for seven days after birth. We investigated the intrarenal expressions of cell cycle regulators p21 and p16 with immunoblots and immunohistochemistry at postnatal day 8. For the determination of renal cellular senescence, immunostaining for senescence-associated ß-galactosidase (SA-ß-gal) and telomerase reverse transcriptase (TERT) was also performed. Enalapril treatment showed significant alterations in cellular senescence in neonatal rat kidneys. In the enalapril-treated group, intrarenal p16 and p21 protein expressions decreased compared to controls. The expressions of both p21 and p16 were reduced throughout the renal cortex and medulla of enalapril-treated rats. The immunoreactivity of TERT in enalapril-treated kidneys was also weaker than that in control kidneys. Control kidneys revealed a clear positive SA-ß-gal signal in the cortical tubules; however, SA-ß-gal activity was noticeably lower in the enalapril-treated kidneys than in control kidneys. Interruption of the RAS during postnatal nephrogenesis may disrupt physiologic renal cellular senescence in the developing rat kidney.


Subject(s)
Angiotensinogen/genetics , Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Kidney/metabolism , p21-Activated Kinases/genetics , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Angiotensins/antagonists & inhibitors , Angiotensins/genetics , Animals , Animals, Newborn/genetics , Animals, Newborn/growth & development , Embryonic Development/genetics , Enalapril/pharmacology , Gene Expression Regulation, Developmental/genetics , Humans , Kidney/growth & development , Kidney Tubules/drug effects , Kidney Tubules/growth & development , Rats , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/genetics , Telomerase/genetics
14.
Int Immunopharmacol ; 89(Pt B): 107081, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33068866

ABSTRACT

The objective of this study was to compare the release of endotoxin and pro-inflammatory cytokines as well as pregnancy outcomes after antibiotic exposure in healthy and bacterial infected pregnant rats. Thirty female Wistar pregnant rats were divided into five groups. Group A considered as control and received intraperitoneal saline 0.9% on 17th day of gestation or DG) and groups B and C treated with 20 mg/kg/day intravenous ceftriaxone and ceftazidime, respectively (DG: 18-20). Groups D and E received intraperitoneal E. coli and LPS on 17th DG respectively. Also, groups F and G received the same treatment as group D but they treated with the exact antibiotics mentioned for groups B and C (same dose and duration). Pregnancy outcomes as well as maternal sera levels of endotoxin, tumor necrosis factor α (TNF-α), interleukin 1ß (IL-1ß), and IL-6 were assessed using enzyme-linked immunosorbent assay. It was shown that group B had a higher IL-1ß (P = 0.003) and TNF-α (P = 0.003) levels compared to the controls (CTC). Group C expressed a lower gestational duration (P = 0.007) as well as higher IL-6 (P = 0.025) and TNF-α (P < 0.001) levels CTC. Interestingly, both group B (P = 0.021) and C (P < 0.001) had a higher rate of endotoxin release CTC. Moreover, in group C, IL-6 (P < 0.0001 and r = -0.941) had a significant correlation with gestational duration. As the results showed, antibiotic administration in non-indication condition seems to be associated with significantly higher production of endotoxin and inflammatory cytokines which increase the risk of poor pregnancy outcomes.


Subject(s)
Anti-Bacterial Agents/adverse effects , Inflammation/etiology , Administration, Intravenous , Animals , Animals, Newborn/genetics , Animals, Newborn/immunology , Anti-Bacterial Agents/administration & dosage , Birth Weight/immunology , Ceftazidime/administration & dosage , Ceftazidime/adverse effects , Ceftriaxone/administration & dosage , Ceftriaxone/adverse effects , Endotoxins/blood , Female , Gene Expression Regulation/immunology , Gestational Age , Interleukin-1beta/blood , Interleukin-6/blood , Pregnancy , Pregnancy Outcome , Rats, Wistar , Tumor Necrosis Factor-alpha/blood
15.
Poult Sci ; 99(10): 4714-4729, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32988506

ABSTRACT

The gut not only plays a key role in digestion and absorption of nutrients but also forms a physical barrier and first line of defense between the host and the luminal environment. A functional gut barrier (mucus and epithelial cells with tight junctions [TJ]) is essential for optimal health and efficient production in poultry. In current broiler system, chicks are deprived of food and water up to 72 h due to uneven hatching, hatchery procedures, and transportation. Post-hatch feed delay results in lower BW, higher FCR and mortality, and delayed post-hatch gut development. Little is known about the effects of early neonatal development and delayed feeding immediately post-hatch on gut barrier function in chickens. Therefore, the aim of the present study was to characterize the expression pattern of gut barrier-related and TJ-related genes in the small intestine of broiler chickens during early development and delay in access to feed. Newly hatched chicks received feed and water immediately after hatch or were subjected to 48 h delayed access to feed to mimic commercial hatchery setting and operations. Birds were sampled (n = 6) at -48, 0, 4, 24, 48, 72, 96, 144, 192, 240, 288, and 336 h post-hatch. Jejunum and ileum were collected, cleaned of digesta, and snap-frozen in liquid nitrogen or fixed in paraformaldehyde. The relative mRNA levels of gut barrier- and TJ-related protein genes were measured by quantitative PCR and analyzed by 2-way ANOVA. In both tissues, changes (P < 0.05) in gene expression pattern of gut barrier-related and TJ-related genes were detected due to delayed access to feed post-hatch and/or development. In general, expression of TJ-related genes was downregulated while mRNA levels of gut barrier-related genes were upregulated during development. Histological differences and changes in mucin staining due to age and treatment were observed. These results suggest that delayed access to feed post-hatch may affect TJ structure and/or function and therefore gut barrier function and overall health of the chicken small intestine.


Subject(s)
Chickens , Feeding Methods , Gene Expression Regulation, Developmental , Intestine, Small , Tight Junctions , Animals , Animals, Newborn/genetics , Chickens/genetics , Feeding Methods/statistics & numerical data , Intestine, Small/metabolism , Tight Junctions/genetics , Time Factors
16.
J Assist Reprod Genet ; 37(11): 2825-2838, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32840762

ABSTRACT

PURPOSE: To keep and increase spermatogonial stem cell number (SSC) is the only available option for pediatric cancer survivors to maintain fertility. Leptin is secreted by the epididymal white adipose tissue and has receptors on stem/progenitor spermatogonia. The purpose of this study is to demonstrate dose- and time-dependent proliferative effect of leptin on stem/progenitor spermatogonia cultures from prepubertal mice testes. METHODS: CD90.2 (+) stem/progenitor spermatogonia were isolated from the C57BL/6 mouse testis on postnatal day 6 and placed in culture. The proliferative effect of leptin supplementation was assessed by colony formation (diameter and number), WST proliferation assays, and xCELLigence real-time cell analysis (RTCA) on days 3, 5, and 7 of culture. Expressions of p-ERK1/2, p-STAT3, total STAT3, and p-SHP2 levels were determined by western blot analysis. RESULTS: Leptin supplementation of 100 ng/ml increased the diameter (p = 0.001) and number (p = 0.01) of colonies in stem/progenitor spermatogonial cultures and caused higher proliferation by WST-1 (p = 0.009) compared with the control on day 7. The EC50 was calculated as 114 ng/ml for leptin by RTCA. Proliferative dose of leptin induced increased expression of p-ERK1/2 (p = 0.009) and p-STAT3 (p = 0.023) on stem/progenitor spermatogonia when compared with the untreated group. CONCLUSION: The results indicated that leptin supplementation exhibited a dose- and time-dependent proliferative effect on stem/progenitor spermatogonia that was associated with increased expression of ERK1/2 and STAT3 pathways while maintaining their undifferentiated state. This output presents a new agent that may help to expand the stem/progenitor spermatogonia pool from the neonatal testis in order to autotransplant after cancer treatment.


Subject(s)
Adult Germline Stem Cells/cytology , Cell Proliferation/genetics , Leptin/genetics , Stem Cells/cytology , Animals , Animals, Newborn/genetics , Animals, Newborn/growth & development , Cell Differentiation/genetics , Humans , Mice
17.
J Dairy Sci ; 103(10): 9515-9524, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32747107

ABSTRACT

The contribution of colostrum to passive immunity transfer and intestinal protection is well known; however, the effects of colostrum intake on the expression of antimicrobial peptides (AP) and Fc receptors in the intestine of neonatal calves are unclear. Our aim was to investigate changes in the expression of AP and Fc receptor in the small intestine of calves in the first 36 h postpartum. Twenty-four Holstein bull calves were used in this study, of which 18 calves were administered 3.2 L of pooled colostrum for each calf per meal via an esophageal tube. Calves were slaughtered at 8 h (1 meal at 1-2 h), 24 h (2 meals at 1-2 h and 10-12 h), and 36 h (3 meals at 1-2 h, 10-12 h, and 22-24 h) postpartum. The remaining 6 calves without any milk administration were slaughtered at 2 h postpartum. Samples of blood and jejunum digesta were collected to determine immunoglobulin concentration using ELISA. Samples of the duodenum, jejunum, and ileum tissues after slaughter were collected to determine AP and Fc receptor expression using quantitative real-time PCR. In calves administered colostrum, IgG concentration in jejunum digesta rapidly decreased in an age-dependent manner (33.41, 9.47, and 0.34 mg/mL at 8, 24, and 36 h, respectively), whereas serum IgG concentration increased significantly, from 0.25 µg/mL at 2 h to 21.72 mg/mL at 24 h. Cathelicidin-4, ß-defensin (DEFB)-7, and enteric ß-defensin expression was upregulated at 8 h postpartum in the duodenum and jejunum compared with that at 2 h, but progressive recovery was detected from 24 h onward. Higher expression of cathelicidin-4, regenerating family member 3γ, lysozyme (LYZ), LYZ1, and LYZ2 and lower expression of DEFB, DEFB1, DEFB7, DEFB10, and enteric ß-defensin were observed in the duodenum and jejunum compared with the ileum. Differences in AP expression between intestinal regions suggested that the innate immune defense mechanism varied significantly among the duodenum, jejunum, and ileum. No difference in the expression of Fc fragment of the IgG receptor was observed either among ages or small intestinal regions. The Fcγ receptor (FcγR)Ia and FcγRIIb expression was the highest at 8 h compared with that at 2, 24, and 36 h, and expression of FcγRIa, FcγRIIb, and FcγRIIIa was higher in the duodenum and jejunum than in the ileum. These results indicated that AP and Fcγ receptors might play important roles in intestinal defense during the passive immunity period.


Subject(s)
Cattle/genetics , Gene Expression , Immunity, Maternally-Acquired/genetics , Pore Forming Cytotoxic Proteins/genetics , Receptors, Fc/genetics , Animals , Animals, Newborn/genetics , Animals, Newborn/immunology , Cattle/immunology , Intestine, Small/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Receptors, Fc/metabolism
18.
Food Funct ; 11(9): 7748-7761, 2020 Sep 23.
Article in English | MEDLINE | ID: mdl-32794529

ABSTRACT

Selenium (Se) is an essential trace element in humans and sows, having a biological function mediated in part by its incorporation into selenoproteins. This study was conducted to investigate the effects of maternal 2-hydroxy-4-methylselenobutanoic acid (HMSeBA), an organic Se source, on reproductive performance, antioxidant capacity and inflammatory status of sows and their offspring. Forty-three Landrace × Yorkshire sows were randomly allocated to receive one of the following three diets during gestation: control diet (control, basal diet, n = 15), sodium selenite (Na2SeO3) supplemented diet (Na2SeO3, basal diet + Na2SeO3 at 0.3 mg Se per kg, n = 13), and HMSeBA supplemented diet (HMSeBA, basal diet + HMSeBA at 0.3 mg Se per kg, n = 15). Blood samples of sows and piglets, placentas and piglet liver samples were analyzed for selenium status, antioxidant capacity and inflammatory cytokines. Results showed that, as compared to the control group, HMSeBA supplementation increased the number of born alive piglets and plasma concentrations of total selenium and selenoprotein P in both sows and piglets. Besides, the activities of antioxidant enzymes in the blood of sows, umbilical cord and piglets, placentas and piglets' liver were increased by dietary HMSeBA supplementation as compared to the control group, while malondialdehyde concentration (p < 0.05) was decreased in the blood of sows, umbilical cord and newborn piglets. In addition, maternal HMSeBA intake during gestation up-regulated antioxidant-related selenoprotein gene expression in the placenta (GPx2, GPx3, p < 0.05) and in the liver of newborn piglets (GPx1, GPx2, GPx3, TXNRD2, p < 0.05). Moreover, as compared to the control group, sows and newborn piglets in the Na2SeO3 and HMSeBA groups had a lower serum interleukin-6 (p < 0.05) concentration, and placentas in the HMSeBA group had lower IL-1ß, IL-6 and IL-8 gene expression (p < 0.05). In conclusion, maternal supplementation of HMSeBA during pregnancy improved antioxidant capacities and reduced the inflammation level in mater, placenta, and fetus. This finding may highlight the important role of selenoproteins (especially GPXs) in preventing negative consequences of over-production of free radicals and inflammatory cytokines during gestation and at births.


Subject(s)
Animals, Newborn/metabolism , Antioxidants/analysis , Butyrates/administration & dosage , Diet/veterinary , Dietary Supplements , Selenium Compounds/administration & dosage , Swine/physiology , Animal Feed , Animal Nutritional Physiological Phenomena , Animals , Animals, Newborn/blood , Animals, Newborn/genetics , Embryo, Mammalian/physiology , Female , Fetal Blood/chemistry , Gene Expression Regulation , Inflammation , Interleukin-1beta/blood , Interleukin-1beta/genetics , Interleukin-6/blood , Interleukin-6/genetics , Oxidation-Reduction , Placenta/chemistry , Pregnancy , Pregnancy Outcome/veterinary , Prenatal Nutritional Physiological Phenomena , Selenium/blood , Selenoprotein P/blood , Swine/embryology , Swine/genetics , Swine/metabolism
19.
Metabolism ; 109: 154296, 2020 08.
Article in English | MEDLINE | ID: mdl-32562799

ABSTRACT

RATIONALE: ApoC2 is an important activator for lipoprotein lipase-mediated hydrolysis of triglyceride-rich plasma lipoproteins. ApoC2-deficient patients display severe hypertriglyceridemia (sHTG) and recurrent acute pancreatitis. However, due to embryonic lethality in ApoC2 deleted mouse extensive understanding of ApoC2 function is limited in mammalian species. OBJECTIVE: We sought to generate an animal model with ApoC2 deficiency in a rodent with some human-like features and then study the precise effects of ApoC2 on lipid and glucose homeostasis. METHODS AND RESULTS: Using CRISPR/Cas9, we deleted Apoc2 gene from golden Syrian hamster and the homozygous (-/-) pups can be born in matured term but exhibited neonatal lethality. By continuous iv administration of normal hamster serum the ApoC2-/- pups could survive till weaning and displayed severe HTG in adulthood on chow diet. A single iv injection of AAV-hApoC2 at birth can also rescue the neonatal death of ApoC2-/- pups. Adult ApoC2-/-hamsters exhibited a unique phenotype of sHTG with hypoglycemia, hypoinsulinemia and spontaneous atherosclerosis. The sHTG in ApoC2-/- adult hamsters could not be corrected by various lipid-lowering medications, but partially ameliorated by medium chain triglyceride diet and completely corrected by AAV-hApoC2. CONCLUSIONS: Our study provides a novel ApoC2-deleted mammalian model with severe hypertriglyceridemia that was fully characterized and highlights a potential therapeutic approach for the treatment of ApoC2 deficient patients.


Subject(s)
Apolipoprotein C-II/deficiency , Atherosclerosis/etiology , Hypertriglyceridemia/etiology , Animals , Animals, Newborn/genetics , Apolipoprotein C-II/therapeutic use , Blood Glucose , Cricetinae , Disease Models, Animal , Gene Knockout Techniques , Homeostasis , Humans , Hypertriglyceridemia/drug therapy , Lipids , Mesocricetus
20.
Mech Dev ; 163: 103616, 2020 09.
Article in English | MEDLINE | ID: mdl-32464196

ABSTRACT

The antagonism between Mdm2 and its close homolog Mdm4 (also known as MdmX) and p53 is vital for embryogenesis and organogenesis. Previously, we demonstrated that targeted disruption of Mdm2 in the Hoxb7+ ureteric bud (Ub) lineage, which gives rise to the renal collecting system, causes renal hypodysplasia culminating in perinatal lethality. In this study, we examine the unique role of Mdm4 in establishing the collecting duct system of the murine kidney. Hoxb7Cre driven loss of Mdm4 in the Ub lineage (UbMdm4-/-) disrupts branching morphogenesis and triggers UB cell apoptosis. UbMdm4-/- kidneys exhibit abnormally dilated Ub tips while the medulla is hypoplastic. These structural alterations result in secondary depletion of nephron progenitors and nascent nephrons. As a result, newborn UbMdm4-/- mice have hypo-dysplastic kidneys. Transcriptional profiling revealed downregulation of the Ret-tyrosine kinase pathway components, Gdnf, Wnt11, Sox8, Etv4 and Cxcr4 in the UbMdm4-/- mice relative to controls. Moreover, the expression levels of the canonical Wnt signaling members Axin2 and Wnt9b are downregulated. Mdm4 deletion upregulated p53 activity and p53-target gene expression including Cdkn1a (p21), Gdf15, Ccng1, PERP, and Fas. Germline loss of p53 in UbMdm4-/- mice largely rescues kidney development and terminal differentiation of the collecting duct. We conclude that Mdm4 plays a unique and vital role in Ub branching morphogenesis and collecting system development.


Subject(s)
Embryonic Development/genetics , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins/genetics , Tumor Suppressor Protein p53/genetics , Animals , Animals, Newborn/genetics , Animals, Newborn/growth & development , Apoptosis/genetics , Axin Protein/genetics , Cell Lineage/genetics , Gene Expression Regulation, Developmental/genetics , Germ Cells/growth & development , Germ Cells/pathology , Homeodomain Proteins/genetics , Kidney/abnormalities , Kidney/metabolism , Mice , Morphogenesis/genetics , Organogenesis/genetics , Ureter/growth & development , Ureter/pathology , Wnt Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...